151
|
Binder Gallimidi A, Fischman S, Revach B, Bulvik R, Maliutina A, Rubinstein AM, Nussbaum G, Elkin M. Periodontal pathogens Porphyromonas gingivalis and Fusobacterium nucleatum promote tumor progression in an oral-specific chemical carcinogenesis model. Oncotarget 2016; 6:22613-23. [PMID: 26158901 PMCID: PMC4673186 DOI: 10.18632/oncotarget.4209] [Citation(s) in RCA: 295] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 05/26/2015] [Indexed: 02/06/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a lethal disease whose incidence is increasing. Epidemiologic studies demonstrate an association between periodontitis and oral cancer, and periodontal pathogens are implicated in the pathogenesis of numerous disorders, including rheumatoid arthritis, cardiovascular diseases, diabetes and gastrointestinal malignancies. Nevertheless, a causal role for periodontal pathogens in OSCC has not been shown, partly due to the lack of an appropriate animal model. Here, utilizing a newly-established murine model of periodontitis-associated oral tumorigenesis, we report that chronic bacterial infection promotes OSCC, and that augmented signaling along the IL-6-STAT3 axis underlies this effect. Our results indicate that periodontal pathogens P. gingivalis and F. nucleatum stimulate tumorigenesis via direct interaction with oral epithelial cells through Toll-like receptors. Furthermore, oral pathogens stimulate human OSCC proliferation and induce expression of key molecules implicated in tumorigenesis. To the best of our knowledge, these findings represent the first demonstration of a mechanistic role for oral bacteria in chemically induced OSCC tumorigenesis. These results are highly relevant for the design of effective prevention and treatment strategies for OSCC.
Collapse
Affiliation(s)
- Adi Binder Gallimidi
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.,Institute of Dental Sciences, Hebrew University-Hadassah Faculty of Dental Medicine, Jerusalem, Israel
| | - Stuart Fischman
- Institute of Dental Sciences, Hebrew University-Hadassah Faculty of Dental Medicine, Jerusalem, Israel
| | - Brurya Revach
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Raanan Bulvik
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Alina Maliutina
- Institute of Dental Sciences, Hebrew University-Hadassah Faculty of Dental Medicine, Jerusalem, Israel
| | - Ariel M Rubinstein
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Gabriel Nussbaum
- Institute of Dental Sciences, Hebrew University-Hadassah Faculty of Dental Medicine, Jerusalem, Israel
| | - Michael Elkin
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
152
|
Hong D, Kurzrock R, Kim Y, Woessner R, Younes A, Nemunaitis J, Fowler N, Zhou T, Schmidt J, Jo M, Lee SJ, Yamashita M, Hughes SG, Fayad L, Piha-Paul S, Nadella MVP, Mohseni M, Lawson D, Reimer C, Blakey DC, Xiao X, Hsu J, Revenko A, Monia BP, MacLeod AR. AZD9150, a next-generation antisense oligonucleotide inhibitor of STAT3 with early evidence of clinical activity in lymphoma and lung cancer. Sci Transl Med 2016; 7:314ra185. [PMID: 26582900 DOI: 10.1126/scitranslmed.aac5272] [Citation(s) in RCA: 355] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Next-generation sequencing technologies have greatly expanded our understanding of cancer genetics. Antisense technology is an attractive platform with the potential to translate these advances into improved cancer therapeutics, because antisense oligonucleotide (ASO) inhibitors can be designed on the basis of gene sequence information alone. Recent human clinical data have demonstrated the potent activity of systemically administered ASOs targeted to genes expressed in the liver. We describe the preclinical activity and initial clinical evaluation of a class of ASOs containing constrained ethyl modifications for targeting the gene encoding the transcription factor STAT3, a notoriously difficult protein to inhibit therapeutically. Systemic delivery of the unformulated ASO, AZD9150, decreased STAT3 expression in a broad range of preclinical cancer models and showed antitumor activity in lymphoma and lung cancer models. AZD9150 preclinical activity translated into single-agent antitumor activity in patients with highly treatment-refractory lymphoma and non-small cell lung cancer in a phase 1 dose-escalation study.
Collapse
Affiliation(s)
- David Hong
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Razelle Kurzrock
- UC San Diego Moores Cancer Center, 3855 Health Sciences Drive, La Jolla, CA 92093, USA.
| | - Youngsoo Kim
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - Richard Woessner
- Cancer Bioscience Drug Discovery, AstraZeneca Pharmaceuticals, 35 Gatehouse Drive, Waltham, MA 02451, USA
| | - Anas Younes
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - John Nemunaitis
- Mary Crowley Cancer Research Center, 7777 Forest Lane, Dallas, TX 75230, USA
| | - Nathan Fowler
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Tianyuan Zhou
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - Joanna Schmidt
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - Minji Jo
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - Samantha J Lee
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - Mason Yamashita
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - Steven G Hughes
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - Luis Fayad
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Sarina Piha-Paul
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Murali V P Nadella
- Drug Safety and Metabolism, AstraZeneca Pharmaceuticals, Waltham, MA 02451, USA
| | - Morvarid Mohseni
- Cancer Bioscience Drug Discovery, AstraZeneca Pharmaceuticals, 35 Gatehouse Drive, Waltham, MA 02451, USA
| | - Deborah Lawson
- Cancer Bioscience Drug Discovery, AstraZeneca Pharmaceuticals, 35 Gatehouse Drive, Waltham, MA 02451, USA
| | - Corinne Reimer
- Cancer Bioscience Drug Discovery, AstraZeneca Pharmaceuticals, 35 Gatehouse Drive, Waltham, MA 02451, USA
| | - David C Blakey
- Oncology iMED, AstraZeneca Pharmaceuticals, Alderley Park, Macclesfield SK10 4TF, UK
| | - Xiaokun Xiao
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - Jeff Hsu
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - Alexey Revenko
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - Brett P Monia
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - A Robert MacLeod
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA.
| |
Collapse
|
153
|
Choudhary MM, France TJ, Teknos TN, Kumar P. Interleukin-6 role in head and neck squamous cell carcinoma progression. World J Otorhinolaryngol Head Neck Surg 2016; 2:90-97. [PMID: 29204553 PMCID: PMC5698512 DOI: 10.1016/j.wjorl.2016.05.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 05/12/2016] [Indexed: 11/04/2022] Open
Abstract
Interleukin-6 (IL-6) is a pleiotropic cytokine which plays an important role in a number of cellular processes including proliferation, survival, differentiation, migration and invasion. IL-6 mediates its downstream effects by activating a number of signaling cascades including JAK/STAT, PI3K/AKT and MAPK pathways. In addition to its effects on tumor cells, IL-6 also regulates tumor progression and tumor metastasis by modulating tumor angiogenesis and tumor lymphangiogenesis. A number of studies have shown that IL-6 levels are markedly upregulated in cancer patients. We and others have shown that high IL-6 expression independently predicts tumor recurrence, tumor metastasis and poor survival in head and neck cancer patients. Therefore targeting IL-6 signaling is a potential therapeutic strategy for the treatment of head and neck squamous cell carcinoma (HNSCC). In this review, we discuss the current understanding of the role of IL-6 in HNSCC progression and potential therapeutic strategies to target IL-6 signaling for the treatment of head and neck cancer patients.
Collapse
Affiliation(s)
- Moaz M Choudhary
- Department of Otolaryngology-Head and Neck Surgery and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Thomas J France
- Department of Otolaryngology-Head and Neck Surgery and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Theodoros N Teknos
- Department of Otolaryngology-Head and Neck Surgery and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Pawan Kumar
- Department of Otolaryngology-Head and Neck Surgery and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
154
|
Peng HY, Cheng YC, Hsu YM, Wu GH, Kuo CC, Liou JP, Chang JY, Jin SLC, Shiah SG. MPT0B098, a Microtubule Inhibitor, Suppresses JAK2/STAT3 Signaling Pathway through Modulation of SOCS3 Stability in Oral Squamous Cell Carcinoma. PLoS One 2016; 11:e0158440. [PMID: 27367272 PMCID: PMC4930189 DOI: 10.1371/journal.pone.0158440] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 06/15/2016] [Indexed: 01/21/2023] Open
Abstract
Microtubule inhibitors have been shown to inhibit Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) signal transduction pathway in various cancer cells. However, little is known of the mechanism by which the microtubule inhibitors inhibit STAT3 activity. In the present study, we examined the effect of a novel small-molecule microtubule inhibitor, MPT0B098, on STAT3 signaling in oral squamous cell carcinoma (OSCC). Treatment of various OSCC cells with MPT0B098 induced growth inhibition, cell cycle arrest and apoptosis, as well as increased the protein level of SOCS3. The accumulation of SOCS3 protein enhanced its binding to JAK2 and TYK2 which facilitated the ubiquitination and degradation of JAK2 and TYK2, resulting in a loss of STAT3 activity. The inhibition of STAT3 activity led to sensitization of OSCC cells to MPT0B098 cytotoxicity, indicating that STAT3 is a key mediator of drug resistance in oral carcinogenesis. Moreover, the combination of MPT0B098 with the clinical drug cisplatin or 5-FU significantly augmented growth inhibition and apoptosis in OSCC cells. Taken together, our results provide a novel mechanism for the action of MPT0B098 in which the JAK2/STAT3 signaling pathway is suppressed through the modulation of SOCS3 protein level. The findings also provide a promising combinational therapy of MPT0B098 for OSCC.
Collapse
Affiliation(s)
- Hsuan-Yu Peng
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
- Department of Life Sciences, National Central University, Taoyuan, Taiwan
| | - Yun-Ching Cheng
- Department of Medical Research, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Yuan-Ming Hsu
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Guan-Hsun Wu
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Ching-Chuan Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Jang-Yang Chang
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
- Division of Hematology and Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medical, National Cheng Kung University, Tainan, Taiwan
| | | | - Shine-Gwo Shiah
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
- * E-mail:
| |
Collapse
|
155
|
Peyser ND, Wang L, Zeng Y, Acquafondata M, Freilino M, Li H, Sen M, Gooding WE, Satake M, Wang Z, Johnson DE, Grandis JR. STAT3 as a Chemoprevention Target in Carcinogen-Induced Head and Neck Squamous Cell Carcinoma. Cancer Prev Res (Phila) 2016; 9:657-63. [PMID: 27267892 DOI: 10.1158/1940-6207.capr-16-0089] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/31/2016] [Indexed: 11/16/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a frequently fatal disease due, in large part, to a high rate of second primary tumor (SPT) formation. The 4-nitroquinoline 1-oxide (4-NQO) mouse model of oral carcinogenesis provides a robust system in which to study chemopreventive agents in the context of chemically induced HNSCC tumors. STAT3 is a potent oncogene that is hyperactivated by tyrosine phosphorylation early in HNSCC carcinogenesis and is a rational therapeutic target. We recently reported that loss-of-function of the STAT3 phosphatase PTPRT promotes STAT3 activation in HNSCC tumors and preclinical models and may serve as a predictive biomarker of response to STAT3 inhibitors, including the small-molecule Stattic. We therefore investigated the hypothesis that Ptprt-knockout (KO) mice would be more susceptible to 4-NQO-induced oral carcinogenesis and more sensitive to Stattic-mediated chemoprevention compared with wild-type (WT) mice. Herein, we demonstrate that Ptprt WT and KO mice develop similar spectra of HNSCC disease severity upon 12 weeks of 4-NQO administration, with no apparent effect of Ptprt genotype on carcinogenesis or treatment outcome. Targeting of STAT3 with Stattic resulted in a chemopreventive effect against 4-NQO-induced oral cancer (P = 0.0402). While these results do not support a central role for PTPRT in 4-NQO-induced HNSCC carcinogenesis, further investigation of STAT3 as a chemoprevention target in this cancer is warranted. Cancer Prev Res; 9(8); 657-63. ©2016 AACR.
Collapse
Affiliation(s)
- Noah D Peyser
- Department of Otolaryngology - Head and Neck Surgery, University of California San Francisco, San Francisco, California
| | - Lin Wang
- Department of Otolaryngology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Yan Zeng
- Department of Otolaryngology - Head and Neck Surgery, University of California San Francisco, San Francisco, California
| | - Marie Acquafondata
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Maria Freilino
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Hua Li
- Department of Otolaryngology - Head and Neck Surgery, University of California San Francisco, San Francisco, California
| | - Malabika Sen
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - William E Gooding
- Department of Otolaryngology and the University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania. Biostatistics Facility, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Masanobu Satake
- Department of Molecular Immunology, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | - Zhenghe Wang
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio. Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Daniel E Johnson
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Jennifer R Grandis
- Department of Otolaryngology - Head and Neck Surgery, University of California San Francisco, San Francisco, California.
| |
Collapse
|
156
|
|
157
|
Wang SJ, Hou YT, Chen LC. A selective decoy-doxorubicin complex for targeted co-delivery, STAT3 probing and synergistic anti-cancer effect. Chem Commun (Camb) 2016. [PMID: 26207858 DOI: 10.1039/c5cc04435a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A novel selective decoy oligodeoxynucleotide (dODN)-doxorubicin (DOX) complex is reported for cancer theranostics. It eliminates the use of a ligand or carrier for targeted delivery and disassembles into therapeutic dODN and DOX upon encountering over-activated STAT3 in cancer cells. Hence, in situ STAT3 probing and synergistic anti-cancer effect are attained at the same time.
Collapse
Affiliation(s)
- Shao-Jen Wang
- Department of Bio-Industrial Mechatronics Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei 10617, Taiwan, Republic of China.
| | | | | |
Collapse
|
158
|
Geinguenaud F, Guenin E, Lalatonne Y, Motte L. Vectorization of Nucleic Acids for Therapeutic Approach: Tutorial Review. ACS Chem Biol 2016; 11:1180-91. [PMID: 26950048 DOI: 10.1021/acschembio.5b01053] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Oligonucleotides present a high therapeutic potential for a wide variety of diseases. However, their clinical development is limited by their degradation by nucleases and their poor blood circulation time. Depending on the administration mode and the cellular target, these macromolecules will have to cross the vascular endothelium, to diffuse through the extracellular matrix, to be transported through the cell membrane, and finally to reach the cytoplasm. To overcome these physiological barriers, many strategies have been developed. Here, we review different methods of DNA vectorization, discuss limitations and advantages of the various vectors, and provide new perspectives for future development.
Collapse
Affiliation(s)
- Frederic Geinguenaud
- Laboratoire CSPBAT,
CNRS UMR 7244, UFR SMBH, Université Paris 13, Sorbonne Paris Cité, F-93017 Bobigny, France
| | - Erwann Guenin
- Inserm, U1148,
Laboratory for Vascular Translational Science, UFR SMBH, Université Paris 13, Sorbonne Paris Cité, F-93017 Bobigny, France
| | - Yoann Lalatonne
- Inserm, U1148,
Laboratory for Vascular Translational Science, UFR SMBH, Université Paris 13, Sorbonne Paris Cité, F-93017 Bobigny, France
- Service
de Médecine Nucléaire, Hôpital Avicenne Assistance Publique-Hôpitaux de Paris 93009 Bobigny France
| | - Laurence Motte
- Inserm, U1148,
Laboratory for Vascular Translational Science, UFR SMBH, Université Paris 13, Sorbonne Paris Cité, F-93017 Bobigny, France
| |
Collapse
|
159
|
Arpin CC, Mac S, Jiang Y, Cheng H, Grimard M, Page BDG, Kamocka MM, Haftchenary S, Su H, Ball DP, Rosa DA, Lai PS, Gómez-Biagi RF, Ali AM, Rana R, Hanenberg H, Kerman K, McElyea KC, Sandusky GE, Gunning PT, Fishel ML. Applying Small Molecule Signal Transducer and Activator of Transcription-3 (STAT3) Protein Inhibitors as Pancreatic Cancer Therapeutics. Mol Cancer Ther 2016; 15:794-805. [PMID: 26873728 PMCID: PMC4873422 DOI: 10.1158/1535-7163.mct-15-0003] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 01/29/2016] [Indexed: 01/02/2023]
Abstract
Constitutively activated STAT3 protein has been found to be a key regulator of pancreatic cancer and a target for molecular therapeutic intervention. In this study, PG-S3-001, a small molecule derived from the SH-4-54 class of STAT3 inhibitors, was found to inhibit patient-derived pancreatic cancer cell proliferation in vitro and in vivo in the low micromolar range. PG-S3-001 binds the STAT3 protein potently, Kd = 324 nmol/L by surface plasmon resonance, and showed no effect in a kinome screen (>100 cancer-relevant kinases). In vitro studies demonstrated potent cell killing as well as inhibition of STAT3 activation in pancreatic cancer cells. To better model the tumor and its microenvironment, we utilized three-dimensional (3D) cultures of patient-derived pancreatic cancer cells in the absence and presence of cancer-associated fibroblasts (CAF). In this coculture model, inhibition of tumor growth is maintained following STAT3 inhibition in the presence of CAFs. Confocal microscopy was used to verify tumor cell death following treatment of 3D cocultures with PG-S3-001. The 3D model was predictive of in vivo efficacy as significant tumor growth inhibition was observed upon administration of PG-S3-001. These studies showed that the inhibition of STAT3 was able to impact the survival of tumor cells in a relevant 3D model, as well as in a xenograft model using patient-derived cells. Mol Cancer Ther; 15(5); 794-805. ©2016 AACR.
Collapse
Affiliation(s)
- Carolyn C Arpin
- Department of Chemistry, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Stephen Mac
- Department of Chemistry, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Yanlin Jiang
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Huiwen Cheng
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Michelle Grimard
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Brent D G Page
- Department of Chemistry, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Malgorzata M Kamocka
- Department of Medicine, Division of Nephrology, Indiana Center for Biological Microscopy, Indiana University School of Medicine, Indianapolis, Indiana
| | - Sina Haftchenary
- Department of Chemistry, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Han Su
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
| | - Daniel P Ball
- Department of Chemistry, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - David A Rosa
- Department of Chemistry, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Ping-Shan Lai
- Department of Chemistry, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Rodolfo F Gómez-Biagi
- Department of Chemistry, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Ahmed M Ali
- Department of Chemistry, University of Toronto Mississauga, Mississauga, Ontario, Canada. Department of Medicinal Chemistry, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Rahul Rana
- Department of Chemistry, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Helmut Hanenberg
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana. Department of Pediatrics III, University Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany. Department of Otorhinolaryngology and Head/Neck Surgery (ENT), Heinrich Heine University, Dusseldorf, Germany
| | - Kagan Kerman
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
| | - Kyle C McElyea
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - George E Sandusky
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Patrick T Gunning
- Department of Chemistry, University of Toronto Mississauga, Mississauga, Ontario, Canada.
| | - Melissa L Fishel
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana. Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
160
|
CADM1 inhibits squamous cell carcinoma progression by reducing STAT3 activity. Sci Rep 2016; 6:24006. [PMID: 27035095 PMCID: PMC4817512 DOI: 10.1038/srep24006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 03/03/2016] [Indexed: 01/11/2023] Open
Abstract
Although squamous cell carcinomas (SqCCs) of the lungs, head and neck, oesophagus, and cervix account for up to 30% of cancer deaths, the mechanisms that regulate disease progression remain incompletely understood. Here, we use gene transduction and human tumor xenograft assays to establish that the tumour suppressor Cell adhesion molecule 1 (CADM1) inhibits SqCC proliferation and invasion, processes fundamental to disease progression. We determine that the extracellular domain of CADM1 mediates these effects by forming a complex with HER2 and integrin α6β4 at the cell surface that disrupts downstream STAT3 activity. We subsequently show that treating CADM1 null tumours with the JAK/STAT inhibitor ruxolitinib mimics CADM1 gene restoration in preventing SqCC growth and metastases. Overall, this study identifies a novel mechanism by which CADM1 prevents SqCC progression and suggests that screening tumours for loss of CADM1 expression will help identify those patients most likely to benefit from JAK/STAT targeted chemotherapies.
Collapse
|
161
|
Klaus M, Prokoph N, Girbig M, Wang X, Huang YH, Srivastava Y, Hou L, Narasimhan K, Kolatkar PR, Francois M, Jauch R. Structure and decoy-mediated inhibition of the SOX18/Prox1-DNA interaction. Nucleic Acids Res 2016; 44:3922-35. [PMID: 26939885 PMCID: PMC4856986 DOI: 10.1093/nar/gkw130] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 02/22/2016] [Indexed: 12/25/2022] Open
Abstract
The transcription factor (TF) SOX18 drives lymphatic vessel development in both embryogenesis and tumour-induced neo-lymphangiogenesis. Genetic disruption of Sox18 in a mouse model protects from tumour metastasis and established the SOX18 protein as a molecular target. Here, we report the crystal structure of the SOX18 DNA binding high-mobility group (HMG) box bound to a DNA element regulating Prox1 transcription. The crystals diffracted to 1.75Å presenting the highest resolution structure of a SOX/DNA complex presently available revealing water structure, structural adjustments at the DNA contact interface and non-canonical conformations of the DNA backbone. To explore alternatives to challenging small molecule approaches for targeting the DNA-binding activity of SOX18, we designed a set of five decoys based on modified Prox1-DNA. Four decoys potently inhibited DNA binding of SOX18 in vitro and did not interact with non-SOX TFs. Serum stability, nuclease resistance and thermal denaturation assays demonstrated that a decoy circularized with a hexaethylene glycol linker and terminal phosphorothioate modifications is most stable. This SOX decoy also interfered with the expression of a luciferase reporter under control of a SOX18-dependent VCAM1 promoter in COS7 cells. Collectively, we propose SOX decoys as potential strategy for inhibiting SOX18 activity to disrupt tumour-induced neo-lymphangiogenesis.
Collapse
Affiliation(s)
- Miriam Klaus
- Genome Regulation Laboratory, Drug Discovery Pipeline, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China Institut für Chemie und Biochemie, Freie Universität Berlin, Thielallee 63, 14195 Berlin, Germany
| | - Nina Prokoph
- Laboratory for Structural Biochemistry, Genome Institute of Singapore, 60 Biopolis Street, 138672 Singapore
| | - Mathias Girbig
- Genome Regulation Laboratory, Drug Discovery Pipeline, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China Institut für Chemie und Biochemie, Freie Universität Berlin, Thielallee 63, 14195 Berlin, Germany
| | - Xuecong Wang
- Genome Regulation Laboratory, Drug Discovery Pipeline, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yong-Heng Huang
- Genome Regulation Laboratory, Drug Discovery Pipeline, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yogesh Srivastava
- Genome Regulation Laboratory, Drug Discovery Pipeline, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Linlin Hou
- Genome Regulation Laboratory, Drug Discovery Pipeline, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Kamesh Narasimhan
- Laboratory for Structural Biochemistry, Genome Institute of Singapore, 60 Biopolis Street, 138672 Singapore
| | - Prasanna R Kolatkar
- Qatar Biomedical Research Institute, Hamad Bin Khalifa Unversity, QatarFoundation, PO Box 5825, Doha, Qatar
| | - Mathias Francois
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Ralf Jauch
- Genome Regulation Laboratory, Drug Discovery Pipeline, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| |
Collapse
|
162
|
Yang R, Rincon M. Mitochondrial Stat3, the Need for Design Thinking. Int J Biol Sci 2016; 12:532-44. [PMID: 27019635 PMCID: PMC4807418 DOI: 10.7150/ijbs.15153] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 02/12/2016] [Indexed: 12/20/2022] Open
Abstract
Stat3 has been studied extensively as a transcription factor, however the finding that Stat3 also localizes to mitochondria has opened a new area to discover non-classical functions. Here we review the current knowledge of mitochondrial Stat3 as a regulator of the electron transport chain (ETC) and its impact on mitochondrial production of ATP and ROS. We also describe recent findings identifying Stat3 as a regulator of mitochondrial Ca(2+) homeostasis through its effect on the ETC. It is becoming evident that these non-classical functions of Stat3 can have a major impact on cancer progression, cardiovascular diseases, and inflammatory diseases. Therefore, mitochondrial Stat3 functions challenge the current design of therapies that solely target Stat3 as a transcription factor and suggest the need for "design thinking," which leads to the development of novel strategies, to intervene the Stat3 pathway.
Collapse
|
163
|
Lee Y, Shin JH, Longmire M, Wang H, Kohrt HE, Chang HY, Sunwoo JB. CD44+ Cells in Head and Neck Squamous Cell Carcinoma Suppress T-Cell-Mediated Immunity by Selective Constitutive and Inducible Expression of PD-L1. Clin Cancer Res 2016; 22:3571-81. [PMID: 26864211 DOI: 10.1158/1078-0432.ccr-15-2665] [Citation(s) in RCA: 165] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 12/28/2015] [Indexed: 12/20/2022]
Abstract
PURPOSE Human tumors consist of heterogeneous populations of cells with distinct marker expression and functional properties. In squamous cell carcinoma of the head and neck (SCCHN), CD44 is a well-characterized marker of a resilient subpopulation of cells associated with increased tumorigenesis, radioresistance, and chemoresistance. Evidence indicates that these cells have an immunosuppressive phenotype; however, mechanisms have been elusive. EXPERIMENTAL DESIGN Using primary human SCCHN tumor samples and patient-derived xenografts, we examined the phenotypes of subsets of tumor cells and investigated mechanisms regulating their immunogenicity. RESULTS CD44(+) cells in primary human SCCHN were found to have an epithelial-to-mesenchymal (EMT) phenotype and were less immunogenic than CD44(-) cells when cultured with autologous CD8(+) tumor-infiltrating T cells. Selective expression of the programmed death-ligand 1 (PD-L1) was observed on CD44(+) cells compared with CD44(-) cells and was associated with constitutive phosphorylation of STAT3 on CD44(+) cells. Importantly, inhibition of STAT3 decreased expression of PD-L1 on CD44(+) cells. IFNγ treatment preferentially induced even further PD-L1 expression on CD44(+) cells and was associated with enhanced IFNγ receptor expression and phosphorylation of STAT1. Finally, the decreased immunogenicity of CD44(+) cells was partially reversed by antibody blockade of the programmed death 1 (PD-1) receptor, indicating that the differences in PD-L1 expression between CD44(+) and CD44(-) cells are biologically and clinically relevant. CONCLUSIONS Our findings provide a mechanism by which long-lived CD44(+) tumor-initiating cells can selectively evade host immune responses and provide rationale for targeting the PD-1 pathway in the adjuvant therapy setting of SCCHN. Clin Cancer Res; 22(14); 3571-81. ©2016 AACR.
Collapse
Affiliation(s)
- Yunqin Lee
- Division of Head and Neck Surgery, Department of Otolaryngology, Stanford Cancer Institute, Stanford, California. Program in Immunology, Stanford University School of Medicine, Stanford, California. Stanford Cancer Institute, Stanford, California. Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - June Ho Shin
- Division of Head and Neck Surgery, Department of Otolaryngology, Stanford Cancer Institute, Stanford, California. Program in Immunology, Stanford University School of Medicine, Stanford, California. Stanford Cancer Institute, Stanford, California. Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Michelle Longmire
- Stanford Cancer Institute, Stanford, California. Program in Epithelial Biology, Stanford Cancer Institute, Stanford, California
| | - Hua Wang
- Stanford Cancer Institute, Stanford, California. Program in Epithelial Biology, Stanford Cancer Institute, Stanford, California
| | - Holbrook E Kohrt
- Program in Immunology, Stanford University School of Medicine, Stanford, California. Stanford Cancer Institute, Stanford, California. Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford, California
| | - Howard Y Chang
- Stanford Cancer Institute, Stanford, California. Program in Epithelial Biology, Stanford Cancer Institute, Stanford, California. Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford, California. Howard Hughes Medical Institute, Stanford, California
| | - John B Sunwoo
- Division of Head and Neck Surgery, Department of Otolaryngology, Stanford Cancer Institute, Stanford, California. Program in Immunology, Stanford University School of Medicine, Stanford, California. Stanford Cancer Institute, Stanford, California. Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
164
|
Serum-resistant CpG-STAT3 decoy for targeting survival and immune checkpoint signaling in acute myeloid leukemia. Blood 2016; 127:1687-700. [PMID: 26796361 DOI: 10.1182/blood-2015-08-665604] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/16/2016] [Indexed: 02/08/2023] Open
Abstract
Targeting oncogenic transcription factor signal transducer and activator of transcription 3 (STAT3) in acute myeloid leukemia (AML) can reduce blast survival and tumor immune evasion. Decoy oligodeoxynucleotides (dODNs), which comprise STAT3-specific DNA sequences are competitive inhibition of STAT3 transcriptional activity. To deliver STAT3dODN specifically to myeloid cells, we linked STAT3dODN to the Toll-like receptor 9 (TLR9) ligand, cytosine guanine dinucleotide (CpG). The CpG-STAT3dODN conjugates are quickly internalized by human and mouse TLR9(+)immune cells (dendritic cells, B cells) and the majority of patients' derived AML blasts, including leukemia stem/progenitor cells. Following uptake, CpG-STAT3dODNs are released from endosomes, and bind and sequester cytoplasmic STAT3, thereby inhibiting downstream gene expression in target cells. STAT3 inhibition in patients' AML cells limits their immunosuppressive potential by reduced arginase expression, thereby partly restoring T-cell proliferation. Partly chemically modified CpG-STAT3dODNs have >60 hours serum half-life which allows for IV administration to leukemia-bearing mice (50% effective dose ∼ 2.5 mg/kg). Repeated administration of CpG-STAT3dODN resulted in regression of human MV4-11 AML in mice. The antitumor efficacy of this strategy is further enhanced in immunocompetent mice by combining direct leukemia-specific cytotoxicity with immunogenic effects of STAT3 blocking/TLR9 triggering. CpG-STAT3dODN effectively reducedCbfb/MYH11/MplAML burden in various organs and eliminated leukemia stem/progenitor cells, mainly through CD8/CD4 T-cell-mediated immune responses. In contrast, small-molecule Janus kinase 2/STAT3 inhibitor failed to reproduce therapeutic effects of cell-selective CpG-STAT3dODN strategy. These results demonstrate therapeutic potential of CpG-STAT3dODN inhibitors with broad implications for treatment of AML and potentially other hematologic malignancies.
Collapse
|
165
|
Abstract
INTRODUCTION This review presents recent developments in the use of nonviral vectors and transfer technologies in cancer gene therapy. Tremendous progress has been made in developing cancer gene therapy in ways that could be applicable to treatments. Numerous efforts are focused on methods of attacking known and novel targets more efficiently and specifically. In parallel to progress in nonviral vector design and delivery technologies, important achievements have been accomplished for suicide, gene replacement, gene suppression and immunostimulatory therapies. New nonviral cancer gene therapies have been developed based on emerging RNAi (si/shRNA-, miRNA) or ODN. AREAS COVERED This review provides an overview of recent gene therapeutic strategies in which nonviral vectors have been used experimentally and in clinical trials. Furthermore, we present current developments in nonviral vector systems in association with important chemical and physical gene delivery technologies and their potential for the future. EXPERT OPINION Nonviral gene therapy has maintained its position as an approach for treating cancer. This is reflected by the fact that more than 17% of all gene therapy trials employ nonviral approaches. Thus, nonviral vectors have emerged as a clinical alternative to viral vectors for the appropriate expression and delivery of therapeutic genes.
Collapse
Affiliation(s)
- Jessica Pahle
- a Experimental and Clinical Research Center , Charité University Medicine Berlin and Max-Delbrück-Center for Moelcular Medicine , Berlin , Germany
| | - Wolfgang Walther
- a Experimental and Clinical Research Center , Charité University Medicine Berlin and Max-Delbrück-Center for Moelcular Medicine , Berlin , Germany
| |
Collapse
|
166
|
Popiolek-Barczyk K, Mika J. Targeting the Microglial Signaling Pathways: New Insights in the Modulation of Neuropathic Pain. Curr Med Chem 2016; 23:2908-2928. [PMID: 27281131 PMCID: PMC5427777 DOI: 10.2174/0929867323666160607120124] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 05/23/2016] [Accepted: 06/06/2016] [Indexed: 12/30/2022]
Abstract
The microglia, once thought only to be supporting cells of the central nervous system (CNS), are now recognized to play essential roles in many pathologies. Many studies within the last decades indicated that the neuro-immune interaction underlies the generation and maintenance of neuropathic pain. Through a large number of receptors and signaling pathways, the microglial cells communicate with neurons, astrocytes and other cells, including those of the immune system. A disturbance or loss of CNS homeostasis causes rapid responses of the microglia, which undergo a multistage activation process. The activated microglia change their cell shapes and gene expression profiles, which induce proliferation, migration, and the production of pro- or antinociceptive factors. The cells release a large number of mediators that can act in a manner detrimental or beneficial to the surrounding cells and can indirectly alter the nociceptive signals. This review discusses the most important microglial intracellular signaling cascades (MAPKs, NF-kB, JAK/STAT, PI3K/Akt) that are essential for neuropathic pain development and maintenance. Our objective was to identify new molecular targets that may result in the development of powerful tools to control the signaling associated with neuropathic pain.
Collapse
Affiliation(s)
| | - Joanna Mika
- Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Str., 31-343 Krakow, Poland.
| |
Collapse
|
167
|
Geiger JL, Grandis JR, Bauman JE. The STAT3 pathway as a therapeutic target in head and neck cancer: Barriers and innovations. Oral Oncol 2015; 56:84-92. [PMID: 26733183 DOI: 10.1016/j.oraloncology.2015.11.022] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 11/12/2015] [Accepted: 11/27/2015] [Indexed: 02/08/2023]
Abstract
Proteins of the signal transducer and activator of transcription (STAT) family mediate cellular responses to cytokines and growth factors. Aberrant regulation of the STAT3 oncogene contributes to tumor formation and progression in many cancers, including head and neck squamous cell carcinoma (HNSCC), where hyperactivation of STAT3 is implicated in both treatment resistance and immune escape. There are no oncogenic gain-of-function mutations in HNSCC. Rather, aberrant STAT3 signaling is primarily driven by upstream growth factor receptors, such as Janus kinase (JAK) and epidermal growth factor receptor (EGFR). Moreover, genomic silencing of select protein tyrosine phosphatase receptors (PTPRs), tumor suppressors that dephosphorylate STAT3, may lead to prolonged phosphorylation and activation of STAT3. This review will summarize current knowledge of the STAT3 pathway and its contribution to HNSCC growth, survival, and resistance to standard therapies, and discuss STAT3-targeting agents in various phases of clinical development.
Collapse
Affiliation(s)
- Jessica L Geiger
- Department of Internal Medicine, Division of Hematology/Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, United States
| | - Jennifer R Grandis
- Department of Otolaryngology, University of California San Francisco, San Francisco, CA, United States
| | - Julie E Bauman
- Department of Internal Medicine, Division of Hematology/Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, United States.
| |
Collapse
|
168
|
Sen M, Pollock NI, Black J, DeGrave KA, Wheeler S, Freilino ML, Joyce S, Lui VWY, Zeng Y, Chiosea SI, Grandis JR. JAK kinase inhibition abrogates STAT3 activation and head and neck squamous cell carcinoma tumor growth. Neoplasia 2015; 17:256-64. [PMID: 25810010 PMCID: PMC4372647 DOI: 10.1016/j.neo.2015.01.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 01/02/2015] [Accepted: 01/07/2015] [Indexed: 12/18/2022] Open
Abstract
Aberrant activation of the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) 3 has been implicated in cell proliferation and survival of many cancers including head and neck squamous cell carcinoma (HNSCC). AZD1480, an orally active pharmacologic inhibitor of JAK1/JAK2, has been tested in several cancer models. In the present study, the in vitro and in vivo effects of AZD1480 were evaluated in HNSCC preclinical models to test the potential use of JAK kinase inhibition for HNSCC therapy. AZD1480 treatment decreased HNSCC proliferation in HNSCC cell lines with half maximal effective concentration (EC50) values ranging from 0.9 to 4 μM in conjunction with reduction of pSTAT3Tyr705 expression. In vivo antitumor efficacy of AZD1480 was demonstrated in patient-derived xenograft (PDX) models derived from two independent HNSCC tumors. Oral administration of AZD1480 reduced tumor growth in conjunction with decreased pSTAT3Tyr705 expression that was observed in both PDX models. These findings suggest that the JAK1/2 inhibitors abrogate STAT3 signaling and may be effective in HNSCC treatment approaches.
Collapse
Affiliation(s)
- Malabika Sen
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Netanya I Pollock
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - John Black
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kara A DeGrave
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sarah Wheeler
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Maria L Freilino
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sonali Joyce
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Vivian W Y Lui
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yan Zeng
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Simion I Chiosea
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jennifer R Grandis
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
169
|
Roeser JC, Leach SD, McAllister F. Emerging strategies for cancer immunoprevention. Oncogene 2015; 34:6029-39. [PMID: 26364615 PMCID: PMC11073473 DOI: 10.1038/onc.2015.98] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/25/2015] [Accepted: 02/27/2015] [Indexed: 12/13/2022]
Abstract
The crucial role of the immune system in the formation and progression of tumors has been widely accepted. On one hand, the surveillance role of the immune system plays an important role in endogenous tumor prevention, but on the other hand, in some special circumstances such as in chronic inflammation, the immune system can actually contribute to the formation and progression of tumors. In recent years, there has been an explosion of novel targeted immunotherapies for advanced cancers. In the present manuscript, we explore known and potential various types of cancer prevention strategies and focus on nonvaccine-based cancer preventive strategies targeting the immune system at the early stages of tumorigenesis.
Collapse
Affiliation(s)
| | - Steven D. Leach
- The David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Florencia McAllister
- Department of Clinical Cancer Prevention. The University of Texas MD Anderson Cancer Center. Houston, TX
| |
Collapse
|
170
|
Yeh JE, Frank DA. STAT3-Interacting Proteins as Modulators of Transcription Factor Function: Implications to Targeted Cancer Therapy. ChemMedChem 2015; 11:795-801. [DOI: 10.1002/cmdc.201500482] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/01/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Jennifer E. Yeh
- Department of Medical Oncology; Dana-Farber Cancer Institute; 450 Brookline Avenue Boston MA 02215 USA
| | - David A. Frank
- Department of Medical Oncology; Dana-Farber Cancer Institute; 450 Brookline Avenue Boston MA 02215 USA
| |
Collapse
|
171
|
Yuan J, Zhang F, Niu R. Multiple regulation pathways and pivotal biological functions of STAT3 in cancer. Sci Rep 2015; 5:17663. [PMID: 26631279 PMCID: PMC4668392 DOI: 10.1038/srep17663] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 11/03/2015] [Indexed: 02/07/2023] Open
Abstract
STAT3 is both a transcription activator and an oncogene that is tightly regulated under normal physiological conditions. However, abundant evidence indicates that STAT3 is persistently activated in several cancers, with a crucial position in tumor onset and progression. In addition to its traditional role in cancer cell proliferation, invasion, and migration, STAT3 also promotes cancer through altering gene expression via epigenetic modification, inducing epithelial–mesenchymal transition (EMT) phenotypes in cancer cells, regulating the tumor microenvironment, and promoting cancer stem cells (CSCs) self-renewal and differentiation. STAT3 is regulated not only by the canonical cytokines and growth factors, but also by the G-protein-coupled receptors, cadherin engagement, Toll-like receptors (TLRs), and microRNA (miRNA). Despite the presence of diverse regulators and pivotal biological functions in cancer, no effective therapeutic inventions are available for inhibiting STAT3 and acquiring potent antitumor effects in the clinic. An improved understanding of the complex roles of STAT3 in cancer is required to achieve optimal therapeutic effects.
Collapse
Affiliation(s)
- Jie Yuan
- Public Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Huan-Hu-Xi Road, Ti-Yuan-Bei, He Xi District, Tianjin, 300060, People's Republic of China
| | - Fei Zhang
- Public Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Huan-Hu-Xi Road, Ti-Yuan-Bei, He Xi District, Tianjin, 300060, People's Republic of China
| | - Ruifang Niu
- Public Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Huan-Hu-Xi Road, Ti-Yuan-Bei, He Xi District, Tianjin, 300060, People's Republic of China
| |
Collapse
|
172
|
Parsel SM, Grandis JR, Thomas SM. Nucleic acid targeting: towards personalized therapy for head and neck cancer. Oncogene 2015; 35:3217-26. [PMID: 26592450 PMCID: PMC4877278 DOI: 10.1038/onc.2015.424] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 09/29/2015] [Accepted: 10/05/2015] [Indexed: 12/13/2022]
Abstract
In light of a detailed characterization of genetic aberrations in cancer, nucleic acid targeting represents an attractive therapeutic approach with significant translational potential. Head and neck squamous cell carcinoma (HNSCC) is a leading cause of cancer deaths worldwide with stagnant 5-year survival rates. Advances in conventional treatment have done little to improve survival and combined chemoradiation is associated with significant adverse effects. Recent reports have characterized the genetic alterations in HNSCC and demonstrated that mutations confer resistance to conventional and molecular targeted therapies. The ability to use specific nucleic acid sequences to inhibit cancer-associated genes including non-druggable targets facilitates personalized medicine approaches with less adverse effects. Additionally, advances in drug delivery mechanisms have increased the transfection efficiency aiding in greater therapeutic responses. Given these advances, the stage has been set to translate the information garnered from genomic studies into personalized treatment strategies. Genes involved in the tumor protein 53 (TP53) and epidermal growth factor receptor (EGFR) pathways have been extensively investigated and many promising preclinical studies have shown tumor inhibition through genetic modulation. We, and others, have demonstrated that targeting oncogene expression with gene therapy approaches is feasible in patients. Other methods such as RNA interference have proven to be effective and are potential candidates for clinical studies. This review summarizes the major advances in sequence-specific gene modulation in the preclinical setting and in clinical trials in head and neck cancer patients.
Collapse
Affiliation(s)
- S M Parsel
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, KS, USA
| | - J R Grandis
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - S M Thomas
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
173
|
Phi JH, Choi SA, Kim SK, Wang KC, Lee JY, Kim DG. Overcoming Chemoresistance of Pediatric Ependymoma by Inhibition of STAT3 Signaling. Transl Oncol 2015; 8:376-386. [PMID: 26500028 PMCID: PMC4631088 DOI: 10.1016/j.tranon.2015.08.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 07/31/2015] [Accepted: 08/10/2015] [Indexed: 01/10/2023] Open
Abstract
The long-term clinical outcome of pediatric intracranial epepdymoma is poor with a high rate of recurrence. One of the main reasons for this poor outcome is the tumor’s inherent resistance to chemotherapy. Signal transducer and activator of transcription 3 (STAT3) is overactive in many human cancers, and inhibition of STAT3 signaling is an emerging area of interest in oncology. In this study, the possibility of STAT3 inhibition as a treatment was investigated in pediatric intracranial ependymoma tissues and cell lines. STAT3 activation status was checked in ependymoma tissues. The responses to conventional chemotherapeutic agents and a STAT3 inhibitor WP1066 in primarily cultured ependymoma cells were measured by cell viability assay. Apoptosis assays were conducted to reveal the cytotoxic mechanism of applied agents. Knockdown of STAT3 was tried to confirm the effects of STAT3 inhibition in ependymoma cells. High levels of phospho-STAT3 (p-STAT3) expression were observed in ependymoma tissue, especially in the anaplastic histology group. There was no cytotoxic effect of cisplatin, ifosfamide, and etoposide. Both brain tumor-initiating cells (BTICs) and bulk tumor cells (BCs) showed considerably decreased viability after WP1066 treatment. However, BTICs had fewer responses than BCs. No additive or synergistic effect was observed for combination therapy of WP1066 and cisplatin. WP1066 effectively abrogated p-STAT3 expression. An increased apoptosis and decreased Survivin expression were observed after WP1066 treatment. Knockdown of STAT3 also decreased cell survival, supporting the critical role of STAT3 in sustaining ependymoma cells. In this study, we observed a cytotoxic effect of STAT3 inhibitor on ependymoma BTICs and BCs. There is urgent need to develop new therapeutic agents for pediatric ependymoma. STAT3 inhibitors may be a new group of drugs for clinical application.
Collapse
Affiliation(s)
- Ji Hoon Phi
- Division of Pediatric Neurosurgery, Seoul National University Children's Hospital, Seoul, Republic of Korea; Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seung-Ah Choi
- Division of Pediatric Neurosurgery, Seoul National University Children's Hospital, Seoul, Republic of Korea; Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seung-Ki Kim
- Division of Pediatric Neurosurgery, Seoul National University Children's Hospital, Seoul, Republic of Korea; Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyu-Chang Wang
- Division of Pediatric Neurosurgery, Seoul National University Children's Hospital, Seoul, Republic of Korea; Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ji Yeoun Lee
- Division of Pediatric Neurosurgery, Seoul National University Children's Hospital, Seoul, Republic of Korea; Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dong Gyu Kim
- Department of Neurosurgery, Seoul National University Hospital, Seoul, Republic of Korea; Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
174
|
Kopechek JA, Carson AR, McTiernan CF, Chen X, Hasjim B, Lavery L, Sen M, Grandis JR, Villanueva FS. Ultrasound Targeted Microbubble Destruction-Mediated Delivery of a Transcription Factor Decoy Inhibits STAT3 Signaling and Tumor Growth. Theranostics 2015; 5:1378-87. [PMID: 26681983 PMCID: PMC4672019 DOI: 10.7150/thno.12822] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 08/19/2015] [Indexed: 01/02/2023] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is constitutively activated in many cancers where it acts to promote tumor progression. A STAT3-specific transcription factor decoy has been developed to suppress STAT3 downstream signaling, but a delivery strategy is needed to improve clinical translation. Ultrasound-targeted microbubble destruction (UTMD) has been shown to enhance image-guided local delivery of molecular therapeutics to a target site. The objective of this study was to deliver STAT3 decoy to squamous cell carcinoma (SCC) tumors using UTMD to disrupt STAT3 signaling and inhibit tumor growth. Studies performed demonstrated that UTMD treatment with STAT3 decoy-loaded microbubbles inhibited STAT3 signaling in SCC cells in vitro. Studies performed in vivo demonstrated that UTMD treatment with STAT3 decoy-loaded microbubbles induced significant tumor growth inhibition (31-51% reduced tumor volume vs. controls, p < 0.05) in mice bearing SCC tumors. Furthermore, expression of STAT3 downstream target genes (Bcl-xL and cyclin D1) was significantly reduced (34-39%, p < 0.05) in tumors receiving UTMD treatment with STAT3 decoy-loaded microbubbles compared to controls. In addition, the quantity of radiolabeled STAT3 decoy detected in tumors eight hours after treatment was significantly higher with UTMD treatment compared to controls (70-150%, p < 0.05). This study demonstrates that UTMD can increase delivery of a transcription factor decoy to tumors in vivo and that the decoy can inhibit STAT3 signaling and tumor growth. These results suggest that UTMD treatment holds potential for clinical use to increase the concentration of a transcription factor signaling inhibitor in the tumor.
Collapse
|
175
|
Iron Oxide Nanoparticles Coated with a Phosphorothioate Oligonucleotide and a Cationic Peptide: Exploring Four Different Ways of Surface Functionalization. NANOMATERIALS 2015; 5:1588-1609. [PMID: 28347083 PMCID: PMC5304778 DOI: 10.3390/nano5041588] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 09/22/2015] [Accepted: 09/23/2015] [Indexed: 01/31/2023]
Abstract
The superparamagnetic iron oxide nanoparticles (SPIONs) have great potential in therapeutic and diagnostic applications. Due to their superparamagnetic behavior, they are used clinically as a Magnetic Resonance Imaging (MRI) contrast agent. Iron oxide nanoparticles are also recognized todays as smart drug-delivery systems. However, to increase their specificity, it is essential to functionalize them with a molecule that effectively targets a specific area of the body. Among the molecules that can fulfill this role, peptides are excellent candidates. Oligonucleotides are recognized as potential drugs for various diseases but suffer from poor uptake and intracellular degradation. In this work, we explore four different strategies, based on the electrostatic interactions between the different partners, to functionalize the surface of SPIONs with a phosphorothioate oligonucleotide (ODN) and a cationic peptide labeled with a fluorophore. The internalization of the nanoparticles has been evaluated in vitro on RAW 264.7 cells. Among these strategies, the "«one-step assembly»", i.e., the direct complexation of oligonucleotides and peptides on iron oxide nanoparticles, provides the best way of coating for the internalization of the nanocomplexes.
Collapse
|
176
|
Lai PS, Rosa DA, Magdy Ali A, Gómez-Biagi RF, Ball DP, Shouksmith AE, Gunning PT. A STAT inhibitor patent review: progress since 2011. Expert Opin Ther Pat 2015; 25:1397-421. [PMID: 26394986 DOI: 10.1517/13543776.2015.1086749] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION The clinical utility of effective direct STAT inhibitors, particularly STAT3 and STAT5, for treating cancer and other diseases is well studied and known. AREAS COVERED This review will highlight the STAT inhibitor patent literature from 2011 to 2015 inclusive. Emphasis will be placed on inhibitors of the STAT3, STAT5a/b, and STAT1 proteins for cancer treatment. The review will, where suitably investigated, describe the mode and the site of inhibition, list indications that were evaluated, and rank the inhibitor's relative potency among compounds in the same class. The reader will gain an understanding of the diverse set of approaches, used both in academia and industry, to target STAT proteins. EXPERT OPINION There is still much work to be done to directly target the STAT3 and STAT5 proteins. As yet, there is still no direct STAT3 inhibitor in the clinic. While the SH2 domain remains a popular target for therapeutic intervention, the DNA-binding domain and N-terminal region are now attracting attention as possible sites for inhibition. Multiple putative STAT3 and STAT5 inhibitors have now been patented across a broad spectrum of chemotypes, each with their own advantages and limitations.
Collapse
Affiliation(s)
- Ping-Shan Lai
- a University of Toronto Mississauga, Department of Chemical and Physical Sciences , 3359 Mississauga Road North, Mississauga, Ontario L5L 1C6, Canada +1 90 55 69 45 88 ; +1 90 55 69 49 29 ;
| | - David A Rosa
- a University of Toronto Mississauga, Department of Chemical and Physical Sciences , 3359 Mississauga Road North, Mississauga, Ontario L5L 1C6, Canada +1 90 55 69 45 88 ; +1 90 55 69 49 29 ;
| | - Ahmed Magdy Ali
- a University of Toronto Mississauga, Department of Chemical and Physical Sciences , 3359 Mississauga Road North, Mississauga, Ontario L5L 1C6, Canada +1 90 55 69 45 88 ; +1 90 55 69 49 29 ;
| | - Rodolfo F Gómez-Biagi
- a University of Toronto Mississauga, Department of Chemical and Physical Sciences , 3359 Mississauga Road North, Mississauga, Ontario L5L 1C6, Canada +1 90 55 69 45 88 ; +1 90 55 69 49 29 ;
| | - Daniel P Ball
- a University of Toronto Mississauga, Department of Chemical and Physical Sciences , 3359 Mississauga Road North, Mississauga, Ontario L5L 1C6, Canada +1 90 55 69 45 88 ; +1 90 55 69 49 29 ;
| | - Andrew E Shouksmith
- a University of Toronto Mississauga, Department of Chemical and Physical Sciences , 3359 Mississauga Road North, Mississauga, Ontario L5L 1C6, Canada +1 90 55 69 45 88 ; +1 90 55 69 49 29 ;
| | - Patrick T Gunning
- a University of Toronto Mississauga, Department of Chemical and Physical Sciences , 3359 Mississauga Road North, Mississauga, Ontario L5L 1C6, Canada +1 90 55 69 45 88 ; +1 90 55 69 49 29 ;
| |
Collapse
|
177
|
Brown ME, Bear MD, Rosol TJ, Premanandan C, Kisseberth WC, London CA. Characterization of STAT3 expression, signaling and inhibition in feline oral squamous cell carcinoma. BMC Vet Res 2015; 11:206. [PMID: 26272737 PMCID: PMC4536595 DOI: 10.1186/s12917-015-0505-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 07/23/2015] [Indexed: 01/19/2023] Open
Abstract
Background Signal transducer and activator of transcription 3 (STAT3) plays a critical role in tumor development by regulating signaling pathways involved in cell proliferation, survival, metastasis and angiogenesis. STAT3 is activated in many cancers, including head and neck squamous cell carcinoma (HNSCC) in people. Feline oral squamous cell carcinoma (OSCC) is similar to advanced or recurrent HNSCC as it is poorly responsive to traditional therapies and carries a poor long-term prognosis. The purpose of this study was to characterize expression and activation of STAT3 in feline OSCC cell lines and tumor samples and to investigate the biologic activity of a novel, allosteric STAT3 inhibitor, LLL12, in feline OSCC cell lines. Results We evaluated 3 feline OSCC cell lines and one of these (SCCF2) exhibited high levels of constitutive STAT3 phosphorylation and high sensitivity to LLL12 treatment. Exposure of SCCF2 cells to LLL12 resulted in decreased expression of pSTAT3 and total STAT3, apoptosis as assessed by caspase 3/7 activation, inhibition of colony formation and reduced expression of the STAT3 transcriptional target survivin. In contrast, the STAT3 transcriptional targets VEGF and MCL-1 increased after LLL12 treatment. This was, in part, likely due to LLL12 mediated upregulation of HIF-1α, which is known to drive VEGF and MCL-1 expression. The OSCC cell lines with low basal STAT3 phosphorylation did not exhibit these effects, suggesting that STAT3 inhibition was responsible for the observed results. Lastly, immunohistochemistry for pSTAT3 was performed using a feline OSCC tissue microarray, demonstrating expression in 48 % of samples tested. Conclusions These data demonstrate that LLL12 has biologic activity against a feline OSCC cell line expressing pSTAT3 and that STAT3 represents a target for therapeutic intervention in this disease. However, given the up-regulation of several STAT3 transcriptional targets following treatment, further investigation of STAT3 and its related signaling pathways in OSCC is warranted. Electronic supplementary material The online version of this article (doi:10.1186/s12917-015-0505-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Megan E Brown
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Misty D Bear
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Thomas J Rosol
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Chris Premanandan
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - William C Kisseberth
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Cheryl A London
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA. .,Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
178
|
Peyser ND, Du Y, Li H, Lui V, Xiao X, Chan TA, Grandis JR. Loss-of-Function PTPRD Mutations Lead to Increased STAT3 Activation and Sensitivity to STAT3 Inhibition in Head and Neck Cancer. PLoS One 2015; 10:e0135750. [PMID: 26267899 PMCID: PMC4534317 DOI: 10.1371/journal.pone.0135750] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 07/25/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Protein tyrosine phosphatase receptor type D (PTPRD) is a putative tumor suppressor in several cancers including head and neck squamous cell carcinoma (HNSCC). STAT3 is a frequently hyperactivated oncogene in HNSCC. As STAT3 is a direct substrate of PTPRD, we sought to determine the genetic or epigenetic alterations of PTPRD that contribute to overactive STAT3 in HNSCC. METHODS We analyzed data from The Cancer Genome Atlas (TCGA) and our previous whole-exome sequencing study and summarized the mutation, methylation, and copy number status of PTPRD in HNSCC and other cancers. In vitro studies involved standard transfection and MTT protocols, as well as methylation-specific PCR. RESULTS Our findings indicate that PTPRD mutation, rather than methylation or copy number alteration, is the primary mechanism by which PTPRD function is lost in HNSCC. We demonstrate that overexpression of wild-type PTPRD in HNSCC cells significantly inhibits growth and STAT3 activation while PTPRD mutants do not, suggesting that mutation may lead to loss of function and subsequent hyper-phosphorylation of PTPRD substrates, especially STAT3. Importantly, we determined that HNSCC cells harboring an endogenous PTPRD mutation are more sensitive to STAT3 blockade than PTPRD wild-type cells. We additionally found that PTPRD mRNA expression does not correlate with pSTAT3 expression, suggesting that alterations that manifest through altered mRNA expression, including hypermethylation and gene copy number alterations, do not significantly contribute to STAT3 overactivation in HNSCC. CONCLUSION PTPRD mutation, but not methylation or copy number loss, may serve as a predictive biomarker of sensitivity to STAT3 inhibitors in HNSCC.
Collapse
Affiliation(s)
- Noah D. Peyser
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America, 15213
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America, 15213
| | - Yu Du
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America, 15213
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America, 15213
- School of Medicine, Tsinghua University, Beijing, China, 100084
| | - Hua Li
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America, 15213
| | - Vivian Lui
- Pharmacogenomics and Precision Therapeutics Laboratory, Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
- Department of Biochemistry, The University of Hong Kong, Hong Kong SAR, China
| | - Xiao Xiao
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America, 15213
| | - Timothy A. Chan
- Human Oncology and Pathogenesis Program and Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, United States of America, 10065
| | - Jennifer R. Grandis
- Department of Otolaryngology, University of California San Francisco, San Francisco, CA, United States of America, 94143
- * E-mail:
| |
Collapse
|
179
|
Yang H, Yamazaki T, Pietrocola F, Zhou H, Zitvogel L, Ma Y, Kroemer G. STAT3 Inhibition Enhances the Therapeutic Efficacy of Immunogenic Chemotherapy by Stimulating Type 1 Interferon Production by Cancer Cells. Cancer Res 2015. [PMID: 26208907 DOI: 10.1158/0008-5472.can-15-1122] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
STAT3 is an oncogenic transcription factor with potent immunosuppressive functions. We found that pharmacologic inhibition of STAT3 or its selective knockout in cancer cells improved the tumor growth-inhibitory efficacy of anthracycline-based chemotherapies. This combined effect of STAT3 inhibition/depletion and anthracyclines was only found in tumors growing on immunocompetent (not in immunodeficient) mice. As compared with Stat3-sufficient control tumors, Stat3(-/-) cancer cells exhibited an increased infiltration by dendritic cells and cytotoxic T lymphocytes after chemotherapy. Anthracyclines are known to induce several stress pathways that enhance the immunogenicity of dying and dead cancer cells, thereby stimulating a dendritic cell-dependent and T lymphocyte-mediated anticancer immune response. Among these therapy-relevant stress pathways, Stat3(-/-) cancer cells manifested one significant improvement, namely an increase in the expression of multiple type-1 interferon-responsive genes, including that of the chemokines Cxcl9 and Cxcl10. This enhanced type-1 interferon response could be suppressed by reintroducing wild-type Stat3 (but not a transactivation-deficient mutant Stat3(Y705F)) into the tumor cells. This maneuver also abolished the improved chemotherapeutic response of Stat3(-/-) cancers. Finally, the neutralization of the common type-1 interferon receptor or that of the chemokine receptor CXCR3 (which binds CXCL9 and CXCL10) abolished the difference in the chemotherapeutic response between Stat3(-/-) and control tumors. Altogether, these results suggest that STAT3 inhibitors may improve the outcome of chemotherapy by enhancing the type-1 interferon response of cancer cells.
Collapse
Affiliation(s)
- Heng Yang
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U 1138, 15 rue de l'Ecole de Médecine, Paris, France. Université Paris Descartes, Sorbonne Paris Cité, 15 rue de l'Ecole de Médecine, Paris, France. Université Pierre et Marie Curie, 15 rue de l'Ecole de Médecine, Paris, France. Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, Villejuif, France. Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China. Suzhou Institute of Systems Medicine, Suzhou, Jiangsu, China
| | - Takahiro Yamazaki
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, Villejuif, France. Institut National de la Santé Et de la Recherche Medicale (INSERM), U1015, GRCC, Villejuif, France. Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 507, Villejuif, France
| | - Federico Pietrocola
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U 1138, 15 rue de l'Ecole de Médecine, Paris, France. Université Paris Descartes, Sorbonne Paris Cité, 15 rue de l'Ecole de Médecine, Paris, France. Université Pierre et Marie Curie, 15 rue de l'Ecole de Médecine, Paris, France. Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, Villejuif, France. University of Paris Sud XI, Kremlin Bicêtre, France
| | - Heng Zhou
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U 1138, 15 rue de l'Ecole de Médecine, Paris, France. Université Paris Descartes, Sorbonne Paris Cité, 15 rue de l'Ecole de Médecine, Paris, France. Université Pierre et Marie Curie, 15 rue de l'Ecole de Médecine, Paris, France. Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, Villejuif, France. University of Paris Sud XI, Kremlin Bicêtre, France
| | - Laurence Zitvogel
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, Villejuif, France. Institut National de la Santé Et de la Recherche Medicale (INSERM), U1015, GRCC, Villejuif, France. Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 507, Villejuif, France. University of Paris Sud XI, Kremlin Bicêtre, France
| | - Yuting Ma
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U 1138, 15 rue de l'Ecole de Médecine, Paris, France. Université Paris Descartes, Sorbonne Paris Cité, 15 rue de l'Ecole de Médecine, Paris, France. Université Pierre et Marie Curie, 15 rue de l'Ecole de Médecine, Paris, France. Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, Villejuif, France. Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China. Suzhou Institute of Systems Medicine, Suzhou, Jiangsu, China.
| | - Guido Kroemer
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U 1138, 15 rue de l'Ecole de Médecine, Paris, France. Université Paris Descartes, Sorbonne Paris Cité, 15 rue de l'Ecole de Médecine, Paris, France. Université Pierre et Marie Curie, 15 rue de l'Ecole de Médecine, Paris, France. Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), 114 rue Edouard Vaillant, Villejuif, France. Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France. Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France. Department of Women's and Children's Health, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
180
|
The role of JAK/STAT signalling in the pathogenesis, prognosis and treatment of solid tumours. Br J Cancer 2015; 113:365-71. [PMID: 26151455 PMCID: PMC4522639 DOI: 10.1038/bjc.2015.233] [Citation(s) in RCA: 434] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 05/17/2015] [Accepted: 05/26/2015] [Indexed: 12/26/2022] Open
Abstract
Aberrant activation of intracellular signalling pathways confers malignant properties on cancer cells. Targeting intracellular signalling pathways has been a productive strategy for drug development, with several drugs acting on signalling pathways already in use and more continually being developed. The JAK/STAT signalling pathway provides an example of this paradigm in haematological malignancies, with the identification of JAK2 mutations in myeloproliferative neoplasms leading to the development of specific clinically effective JAK2 inhibitors, such as ruxolitinib. It is now clear that many solid tumours also show activation of JAK/STAT signalling. In this review, we focus on the role of JAK/STAT signalling in solid tumours, examining the molecular mechanisms that cause inappropriate pathway activation and their cellular consequences. We also discuss the degree to which activated JAK/STAT signalling contributes to oncogenesis. Studies showing the effect of activation of JAK/STAT signalling upon prognosis in several tumour types are summarised. Finally, we discuss the prospects for treating solid tumours using strategies targeting JAK/STAT signalling, including what can be learned from haematological malignancies and the extent to which results in solid tumours might be expected to differ.
Collapse
|
181
|
Gaykalova DA, Manola JB, Ozawa H, Zizkova V, Morton K, Bishop JA, Sharma R, Zhang C, Michailidi C, Considine M, Tan M, Fertig EJ, Hennessey PT, Ahn J, Koch WM, Westra WH, Khan Z, Chung CH, Ochs MF, Califano JA. NF-κB and stat3 transcription factor signatures differentiate HPV-positive and HPV-negative head and neck squamous cell carcinoma. Int J Cancer 2015; 137:1879-89. [PMID: 25857630 DOI: 10.1002/ijc.29558] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 03/27/2015] [Indexed: 12/29/2022]
Abstract
Using high-throughput analyses and the TRANSFAC database, we characterized TF signatures of head and neck squamous cell carcinoma (HNSCC) subgroups by inferential analysis of target gene expression, correcting for the effects of DNA methylation and copy number. Using this discovery pipeline, we determined that human papillomavirus-related (HPV+) and HPV- HNSCC differed significantly based on the activity levels of key TFs including AP1, STATs, NF-κB and p53. Immunohistochemical analysis confirmed that HPV- HNSCC is characterized by co-activated STAT3 and NF-κB pathways and functional studies demonstrate that this phenotype can be effectively targeted with combined anti-NF-κB and anti-STAT therapies. These discoveries correlate strongly with previous findings connecting STATs, NF-κB and AP1 in HNSCC. We identified five top-scoring pair biomarkers from STATs, NF-κB and AP1 pathways that distinguish HPV+ from HPV- HNSCC based on TF activity and validated these biomarkers on TCGA and on independent validation cohorts. We conclude that a novel approach to TF pathway analysis can provide insight into therapeutic targeting of patient subgroup for heterogeneous disease such as HNSCC.
Collapse
Affiliation(s)
- Daria A Gaykalova
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Judith B Manola
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA
| | - Hiroyuki Ozawa
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Veronika Zizkova
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, MD.,Laboratory of Molecular Pathology, Institute of Molecular and Translational Medicine, Palacky University, Olomouc, Czech Republic
| | - Kathryn Morton
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Justin A Bishop
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, MD.,Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Rajni Sharma
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Chi Zhang
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, MD.,University of Virginia, Charlottesville, VA
| | - Christina Michailidi
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Michael Considine
- Division of Oncology Biostatistics, Johns Hopkins Medical Institutions, Baltimore, MD.,Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Marietta Tan
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Elana J Fertig
- Division of Oncology Biostatistics, Johns Hopkins Medical Institutions, Baltimore, MD.,Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Patrick T Hennessey
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, MD.,Mid-Michigan Ear, Nose, and Throat, East Lansing, MI
| | - Julie Ahn
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Wayne M Koch
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, MD
| | - William H Westra
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, MD.,Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Zubair Khan
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Christine H Chung
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, MD.,Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Michael F Ochs
- Division of Oncology Biostatistics, Johns Hopkins Medical Institutions, Baltimore, MD.,Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD.,Department of Mathematics and Statistics, The College of New Jersey, Ewing, NJ
| | - Joseph A Califano
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, MD.,Milton J. Dance Head and Neck Center, Greater Baltimore Medical Center, Baltimore, MD
| |
Collapse
|
182
|
Balmert SC, Zmolek AC, Glowacki AJ, Knab TD, Rothstein SN, Wokpetah JM, Fedorchak MV, Little SR. Positive Charge of "Sticky" Peptides and Proteins Impedes Release From Negatively Charged PLGA Matrices. J Mater Chem B 2015; 3:4723-4734. [PMID: 26085928 PMCID: PMC4465798 DOI: 10.1039/c5tb00515a] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The influence of electrostatic interactions and/or acylation on release of charged ("sticky") agents from biodegradable polymer matrices was systematically characterized. We hypothesized that release of peptides with positive charge would be hindered from negatively charged poly(lactic-co-glycolic acid) (PLGA) microparticles. Thus, we investigated release of peptides with different degrees of positive charge from several PLGA microparticle formulations, with different molecular weights and/or end groups (acid- or ester-terminated). Indeed, release studies revealed distinct inverse correlations between the amount of positive charge on peptides and their release rates from each PLGA microparticle formulation. Furthermore, we examined the case of peptides with net charge that changes from negative to positive within the pH range observed in degrading microparticles. These charge changing peptides displayed counterintuitive release kinetics, initially releasing faster from slower degrading (less acidic) microparticles, and releasing slower from the faster degrading (more acidic) microparticles. Importantly, trends between agent charge and release rates for model peptides also translated to larger, therapeutically relevant proteins and oligonucleotides. The results of these studies may improve future design of controlled release systems for numerous therapeutic biomolecules exhibiting positive charge, ultimately reducing time-consuming and costly trial and error iterations of such formulations.
Collapse
Affiliation(s)
- Stephen C. Balmert
- Department of Bioengineering, University of Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, USA
| | - Andrew C. Zmolek
- Department of Chemical Engineering, University of Pittsburgh, PA, USA
| | - Andrew J. Glowacki
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, USA
- Department of Chemical Engineering, University of Pittsburgh, PA, USA
| | - Timothy D. Knab
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, USA
- Department of Chemical Engineering, University of Pittsburgh, PA, USA
| | - Sam N. Rothstein
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, USA
- Department of Chemical Engineering, University of Pittsburgh, PA, USA
| | | | - Morgan V. Fedorchak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, USA
- Department of Chemical Engineering, University of Pittsburgh, PA, USA
- Department of Ophthalmology, University of Pittsburgh, PA, USA
| | - Steven R. Little
- Department of Bioengineering, University of Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, USA
- Department of Chemical Engineering, University of Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, PA, USA
| |
Collapse
|
183
|
Wake MS, Watson CJ. STAT3 the oncogene - still eluding therapy? FEBS J 2015; 282:2600-11. [DOI: 10.1111/febs.13285] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 03/04/2015] [Accepted: 03/26/2015] [Indexed: 02/06/2023]
|
184
|
Benveniste EN, Liu Y, McFarland BC, Qin H. Involvement of the janus kinase/signal transducer and activator of transcription signaling pathway in multiple sclerosis and the animal model of experimental autoimmune encephalomyelitis. J Interferon Cytokine Res 2015; 34:577-88. [PMID: 25084174 DOI: 10.1089/jir.2014.0012] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Multiple sclerosis (MS) and its animal model of experimental autoimmune encephalomyelitis (EAE) are characterized by focal inflammatory infiltrates into the central nervous system, demyelinating lesions, axonal damage, and abundant production of cytokines that activate immune cells and damage neurons and oligodendrocytes, including interleukin-12 (IL-12), IL-6, IL-17, IL-21, IL-23, granulocyte macrophage-colony stimulating factor, and interferon-gamma. The Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) signaling pathway mediates the biological activities of these cytokines and is essential for the development and regulation of immune responses. Dysregulation of the JAK/STAT pathway contributes to numerous autoimmune diseases, including MS/EAE. The JAK/STAT pathway is aberrantly activated in MS/EAE because of excessive production of cytokines, loss of expression of negative regulators such as suppressors of cytokine signaling proteins, and significant enrichment of genes encoding components of the JAK/STAT pathway, including STAT3. Specific JAK/STAT inhibitors have been used in numerous preclinical models of MS and demonstrate beneficial effects on the clinical course of disease and attenuation of innate and adaptive immune responses. In addition, other drugs such as statins, glatiramer acetate, laquinimod, and fumarates have beneficial effects that involve inhibition of the JAK/STAT pathway. We conclude by discussing the feasibility of the JAK/STAT pathway as a target for neuroinflammatory diseases.
Collapse
Affiliation(s)
- Etty N Benveniste
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham , Birmingham, Alabama
| | | | | | | |
Collapse
|
185
|
Johnston SJ, Carroll JS. Transcription factors and chromatin proteins as therapeutic targets in cancer. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1855:183-92. [PMID: 25721328 DOI: 10.1016/j.bbcan.2015.02.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/09/2015] [Accepted: 02/09/2015] [Indexed: 01/15/2023]
Abstract
Targeting the factors that regulate gene transcription is a compelling strategy in cancer therapeutics. Traditionally, these have been considered intractable targets, but recent work has revealed novel strategies for the regulation of transcription factor activity in cancer. This review will highlight some of the emerging concepts and provide examples where agents that target transcription factors are being exploited clinically for cancer therapies.
Collapse
Affiliation(s)
- Simon J Johnston
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UK
| | - Jason S Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UK.
| |
Collapse
|
186
|
Rath T, Billmeier U, Waldner MJ, Atreya R, Neurath MF. From physiology to disease and targeted therapy: interleukin-6 in inflammation and inflammation-associated carcinogenesis. Arch Toxicol 2015; 89:541-554. [PMID: 25632846 DOI: 10.1007/s00204-015-1461-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/08/2015] [Indexed: 12/29/2022]
Abstract
Since its discovery in 1986, originally as B cell stimulating factor 2, the knowledge on IL-6 for immune homeostasis and its pathophysiological implications has rapidly increased. It is now clear that IL-6, alone or in combination with other cytokines, is an architect for shaping and generating immune responses which exerts profound activities on the induction of acute-phase reactions, the differentiation of B lymphocytes, the modulation of T cell apoptosis, the activation of T helper cells and the balance between regulatory T cells and Th17 cells. In parallel to the identification of these physiologic functions, IL-6 has emerged as a critical mediator for perpetuating chronic inflammation and autoimmunity and is increasingly recognized as a key cytokine for linking chronic inflammation to cancer development. In this review, we begin by briefly summarizing the molecular events of IL-6 regulation and signaling and then describe the role of IL-6 in orchestrating innate and adaptive immune responses and its immunopathological relevance for chronic inflammatory diseases. We further outline how IL-6 links chronic inflammation and cancer development and finally provide an outlook on novel therapeutic strategies targeting IL-6 signaling for the treatment of chronic inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Timo Rath
- Division of Gastroenterology, Department of Medicine 1, University Hospital Erlangen, FAU Erlangen-Nuremberg, Ulmenweg 18, 91054, Erlangen, Germany,
| | | | | | | | | |
Collapse
|
187
|
Gritsina G, Xiao F, O'Brien SW, Gabbasov R, Maglaty MA, Xu RH, Thapa RJ, Zhou Y, Nicolas E, Litwin S, Balachandran S, Sigal LJ, Huszar D, Connolly DC. Targeted Blockade of JAK/STAT3 Signaling Inhibits Ovarian Carcinoma Growth. Mol Cancer Ther 2015; 14:1035-47. [PMID: 25646015 PMCID: PMC4394029 DOI: 10.1158/1535-7163.mct-14-0800] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 01/26/2015] [Indexed: 12/22/2022]
Abstract
Ovarian carcinoma is the fifth leading cause of death among women in the United States. Persistent activation of STAT3 is frequently detected in ovarian carcinoma. STAT3 is activated by Janus family kinases (JAK) via cytokine receptors, growth factor receptor, and non-growth factor receptor tyrosine kinases. Activation of STAT3 mediates tumor cell proliferation, survival, motility, invasion, and angiogenesis, and recent work demonstrates that STAT3 activation suppresses antitumor immune responses and supports tumor-promoting inflammation. We hypothesized that therapeutic targeting of the JAK/STAT3 pathway would inhibit tumor growth by direct effects on ovarian carcinoma cells and by inhibition of cells in the tumor microenvironment (TME). To test this, we evaluated the effects of a small-molecule JAK inhibitor, AZD1480, on cell viability, apoptosis, proliferation, migration, and adhesion of ovarian carcinoma cells in vitro. We then evaluated the effects of AZD1480 on in vivo tumor growth and progression, gene expression, tumor-associated matrix metalloproteinase (MMP) activity, and immune cell populations in a transgenic mouse model of ovarian carcinoma. AZD1480 treatment inhibited STAT3 phosphorylation and DNA binding, and migration and adhesion of cultured ovarian carcinoma cells and ovarian tumor growth rate, volume, and ascites production in mice. In addition, drug treatment led to altered gene expression, decreased tumor-associated MMP activity, and fewer suppressor T cells in the peritoneal TME of tumor-bearing mice than control mice. Taken together, our results show pharmacologic inhibition of the JAK2/STAT3 pathway leads to disruption of functions essential for ovarian tumor growth and progression and represents a promising therapeutic strategy.
Collapse
Affiliation(s)
- Galina Gritsina
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Fang Xiao
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Shane W O'Brien
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Rashid Gabbasov
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania. Kazan (Volga Region) Federal University, Kazan, Russia
| | - Marisa A Maglaty
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Ren-Huan Xu
- Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Roshan J Thapa
- Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Yan Zhou
- Biostatistics Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | | | - Samuel Litwin
- Biostatistics Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Siddharth Balachandran
- Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Luis J Sigal
- Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | | | - Denise C Connolly
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.
| |
Collapse
|
188
|
Jiang Y, Ludwig J, Janku F. Targeted therapies for advanced Ewing sarcoma family of tumors. Cancer Treat Rev 2015; 41:391-400. [PMID: 25869102 DOI: 10.1016/j.ctrv.2015.03.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/17/2015] [Accepted: 03/20/2015] [Indexed: 12/30/2022]
Abstract
The prognosis of adolescent and young adult patients battling metastatic Ewing sarcoma family of tumors (ESFT) remains less than 30% despite the development of systemic therapies. In the era of personalized medicine, novel molecular targets have been tested in preclinical or clinical settings in ESFT. In this review, we focus on early clinical and translational research that identified multiple molecular targets, including IGF-1R; mTOR; tyrosine kinase inhibitors; EWS-FLI1-related targets, and others. Overall, novel targeted therapies demonstrated modest efficacy; however pronounced and durable antineoplastic responses have been observed in small subsets of treated patients, for example with IGF-1R antibodies. Identifying outcome-predicting biomarkers and overcoming treatment resistance remain major challenges. Due to the rarity of ESFT, multi-institutional collaboration efforts of clinicians, basic and translational scientists are needed in order to understand biology of therapeutic response or resistance, which can lead to development of novel therapeutic methods and improved patient outcomes.
Collapse
Affiliation(s)
- Yunyun Jiang
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph Ludwig
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Filip Janku
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
189
|
Auphan-Anezin N, Schmitt-Verhulst AM. Silence STAT3 in the procancer niche… and activate CD8+ T cells to kill premetastatic myeloid intruders. Eur J Immunol 2015; 45:44-8. [PMID: 25471823 DOI: 10.1002/eji.201445300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 11/25/2014] [Accepted: 11/28/2014] [Indexed: 12/27/2022]
Abstract
Several recent studies have implicated myeloid cells in providing a microenvironment that promotes tumor cell survival and metastasis, therefore preparing a "premetastatic niche" for cancer progression. In this issue of the European Journal of Immunology, Zhang et al. [Eur. J. Immunol. 2015. 45: 71-81] address the regulation of immune cells in premetastatic lymph nodes in experimental mouse models. The authors show that signal transducer and activator of transcription 3 (STAT3) ablation in murine myeloid cells, which renders the premetastatic niche less receptive to metastasis by B16 melanoma cells, also leads to local activation in the niche of CD8(+) T cells with increased expression of IFN-γ and granzyme B. Data further suggest that STAT3 activation in the myeloid population leads to poor tumor antigen presenting capacity as well as resistance to CD8(+) T-cell killing. Based on these studies in mice and observations in human cancer patients, the authors propose treatments designed to regulate STAT3 activation, which are correlated with increased cytolytic activity of CD8(+) T cells in mouse models.
Collapse
Affiliation(s)
- Nathalie Auphan-Anezin
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University UM2, Marseille, France; INSERM UMR 1104, Marseille, France; CNRS UMR 7280, Marseille, France
| | | |
Collapse
|
190
|
Dutzmann J, Daniel JM, Bauersachs J, Hilfiker-Kleiner D, Sedding DG. Emerging translational approaches to target STAT3 signalling and its impact on vascular disease. Cardiovasc Res 2015; 106:365-74. [PMID: 25784694 PMCID: PMC4431663 DOI: 10.1093/cvr/cvv103] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 02/05/2015] [Indexed: 12/30/2022] Open
Abstract
Acute and chronic inflammation responses characterize the vascular remodelling processes in atherosclerosis, restenosis, pulmonary arterial hypertension, and angiogenesis. The functional and phenotypic changes in diverse vascular cell types are mediated by complex signalling cascades that initiate and control genetic reprogramming. The signalling molecule's signal transducer and activator of transcription 3 (STAT3) plays a key role in the initiation and continuation of these pathophysiological changes. This review highlights the pivotal involvement of STAT3 in pathological vascular remodelling processes and discusses potential translational therapies, which target STAT3 signalling, to prevent and treat cardiovascular diseases. Moreover, current clinical trials using highly effective and selective inhibitors of STAT3 signalling for distinct diseases, such as myelofibrosis and rheumatoid arthritis, are discussed with regard to their vascular (side-) effects and their potential to pave the way for a direct use of these molecules for the prevention or treatment of vascular diseases.
Collapse
Affiliation(s)
- Jochen Dutzmann
- Vascular Remodeling and Regeneration Group, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover 30625, Germany
| | - Jan-Marcus Daniel
- Vascular Remodeling and Regeneration Group, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover 30625, Germany
| | - Johann Bauersachs
- Vascular Remodeling and Regeneration Group, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover 30625, Germany
| | - Denise Hilfiker-Kleiner
- Vascular Remodeling and Regeneration Group, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover 30625, Germany
| | - Daniel G Sedding
- Vascular Remodeling and Regeneration Group, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover 30625, Germany
| |
Collapse
|
191
|
Feber A, Arya M, de Winter P, Saqib M, Nigam R, Malone PR, Tan WS, Rodney S, Lechner M, Freeman A, Jameson C, Muneer A, Beck S, Kelly JD. Epigenetics markers of metastasis and HPV-induced tumorigenesis in penile cancer. Clin Cancer Res 2015; 21:1196-206. [PMID: 25538261 PMCID: PMC4351916 DOI: 10.1158/1078-0432.ccr-14-1656] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE Penile cancer is a rare malignancy in the developed world with just more than 1,600 new cases diagnosed in the United States per year; however, the incidence is much higher in developing countries. Although HPV is known to contribute to tumorigenesis, little is known about the genetic or epigenetic alterations defining penile cancer. EXPERIMENTAL DESIGN Using high-density genome-wide methylation arrays, we have identified epigenetic alterations associated with penile cancer. Q-MSP was used to validate lymph node metastasis markers in 50 cases. A total of 446 head and neck squamous cell carcinoma (HNSCC) and cervical squamous cell carcinoma (CESCC) samples were used to validate HPV-associated epigenetic alterations. RESULTS We defined 6,933 methylation variable positions (MVP) between normal and tumor tissue, which includes 997 hypermethylated differentially methylated regions associated with tumor supressor genes, including CDO1, AR1, and WT1. Analysis of penile cancer tumors identified a 4 gene epi-signature which accurately predicted lymph node metastasis in an independent cohort (AUC of 89%). Finally, we explored the epigenetic alterations associated with penile cancer HPV infection and defined a 30 loci lineage-independent HPV specific epi-signature which predicts HPV status and survival in independent HNSCC, CESC cohorts. Epi-signature-negative patients have a significantly worse overall survival [HNSCC P = 0.00073; 95% confidence interval (CI), 0.021-0.78; CESC P = 0.0094; HR = 3.91, 95% CI = 0.13-0.78], HPV epi-signature is a better predictor of survival than HPV status alone. CONCLUSIONS These data demonstrate for the first time genome-wide epigenetic events involved in an aggressive penile cancer phenotype and define the epigenetic alterations common across multiple HPV-driven malignancies.
Collapse
Affiliation(s)
- Andrew Feber
- UCL Cancer Institute, University College London, London, United Kingdom.
| | - Manit Arya
- Division of Surgery and Interventional Science, UCL Medical School, University College London, London, United Kingdom. Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Patricia de Winter
- Division of Surgery and Interventional Science, UCL Medical School, University College London, London, United Kingdom
| | - Muhammad Saqib
- Division of Surgery and Interventional Science, UCL Medical School, University College London, London, United Kingdom
| | - Raj Nigam
- University College London Hospital, 250 Euston Road, London, United Kingdom
| | - Peter R Malone
- University College London Hospital, 250 Euston Road, London, United Kingdom
| | - Wei Shen Tan
- Division of Surgery and Interventional Science, UCL Medical School, University College London, London, United Kingdom
| | - Simon Rodney
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Matthias Lechner
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Alex Freeman
- Department of Histopathology, University College London Hospital, London, United Kingdom
| | - Charles Jameson
- Department of Histopathology, University College London Hospital, London, United Kingdom
| | - Asif Muneer
- Division of Surgery and Interventional Science, UCL Medical School, University College London, London, United Kingdom. University College London Hospital, 250 Euston Road, London, United Kingdom
| | - Stephan Beck
- UCL Cancer Institute, University College London, London, United Kingdom
| | - John D Kelly
- UCL Cancer Institute, University College London, London, United Kingdom. Division of Surgery and Interventional Science, UCL Medical School, University College London, London, United Kingdom.
| |
Collapse
|
192
|
Rath T, Billmeier U, Waldner MJ, Atreya R, Neurath MF. From physiology to disease and targeted therapy: interleukin-6 in inflammation and inflammation-associated carcinogenesis. Arch Toxicol 2015. [DOI: 10.1007/s00204-015-1461-5 and 3010=3010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
193
|
From physiology to disease and targeted therapy: interleukin-6 in inflammation and inflammation-associated carcinogenesis. Arch Toxicol 2015. [DOI: 10.1007/s00204-015-1461-5 waitfor delay '0:0:5'-- ismb] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
194
|
Rath T, Billmeier U, Waldner MJ, Atreya R, Neurath MF. From physiology to disease and targeted therapy: interleukin-6 in inflammation and inflammation-associated carcinogenesis. Arch Toxicol 2015. [DOI: 10.1007/s00204-015-1461-5 and (select 9484 from(select count(*),concat(0x716b627871,(select (elt(9484=9484,1))),0x716a787671,floor(rand(0)*2))x from information_schema.plugins group by x)a)] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
|
195
|
Rath T, Billmeier U, Waldner MJ, Atreya R, Neurath MF. From physiology to disease and targeted therapy: interleukin-6 in inflammation and inflammation-associated carcinogenesis. Arch Toxicol 2015. [DOI: 10.1007/s00204-015-1461-5 order by 1-- kwdt] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
196
|
|
197
|
From physiology to disease and targeted therapy: interleukin-6 in inflammation and inflammation-associated carcinogenesis. Arch Toxicol 2015. [DOI: 10.1007/s00204-015-1461-5 waitfor delay '0:0:5'] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
|
198
|
From physiology to disease and targeted therapy: interleukin-6 in inflammation and inflammation-associated carcinogenesis. Arch Toxicol 2015. [DOI: 10.1007/s00204-015-1461-5 order by 1-- esve] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
|
199
|
Rath T, Billmeier U, Waldner MJ, Atreya R, Neurath MF. From physiology to disease and targeted therapy: interleukin-6 in inflammation and inflammation-associated carcinogenesis. Arch Toxicol 2015. [DOI: 10.1007/s00204-015-1461-5 and 3010=3010-- kvwx] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
200
|
Rath T, Billmeier U, Waldner MJ, Atreya R, Neurath MF. From physiology to disease and targeted therapy: interleukin-6 in inflammation and inflammation-associated carcinogenesis. Arch Toxicol 2015. [DOI: 10.1007/s00204-015-1461-5 and 5674=dbms_pipe.receive_message(chr(81)||chr(112)||chr(90)||chr(102),5)] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
|